Abstract

IntroductionPoor translation of efficacy data derived from animal models can lead to clinical trials unlikely to benefit patients–or even put them at risk–and is a potential contributor to costly and unnecessary attrition in drug development.ObjectivesTo develop a tool to assess, validate and compare the clinical translatability of animal models used for the preliminary assessment of efficacy.Design and resultsWe performed a scoping review to identify the key aspects used to validate animal models. Eight domains (Epidemiology, Symptomatology and Natural History–SNH, Genetic, Biochemistry, Aetiology, Histology, Pharmacology and Endpoints) were identified. We drafted questions to evaluate the different facets of human disease simulation. We designed the Framework to Identify Models of Disease (FIMD) to include standardised instructions, a weighting and scoring system to compare models as well as factors to help interpret model similarity and evidence uncertainty. We also added a reporting quality and risk of bias assessment of drug intervention studies in the Pharmacological Validation domain. A web-based survey was conducted with experts from different stakeholders to gather input on the framework. We conducted a pilot study of the validation in two models for Type 2 Diabetes (T2D)–the ZDF rat and db/db mouse. Finally, we present a full validation and comparison of two animal models for Duchenne Muscular Dystrophy (DMD): the mdx mouse and GRMD dog. We show that there are significant differences between the mdx mouse and the GRMD dog, the latter mimicking the human epidemiological, SNH, and histological aspects to a greater extent than the mouse despite the overall lack of published data.ConclusionsFIMD facilitates drug development by serving as the basis to select the most relevant model that can provide meaningful data and is more likely to generate translatable results to progress drug candidates to the clinic.

Highlights

  • Poor translation of efficacy data derived from animal models can lead to clinical trials unlikely to benefit patients–or even put them at risk–and is a potential contributor to costly and unnecessary attrition in drug development

  • The use of non-human animals to evaluate the safety of new drugs is an integral part of the regulatory research and development process [1,2]

  • Despite their apparent value as a drug testing system to predict safety and efficacy in humans, scientists are increasingly aware of their considerable drawbacks and limited predictivity [3,4,5,6]

Read more

Summary

Objectives

To develop a tool to assess, validate and compare the clinical translatability of animal models used for the preliminary assessment of efficacy. Data Availability Statement: All relevant data are within the manuscript and its Supporting Information files. We performed a scoping review to identify the key aspects used to validate animal models. We conducted a pilot study of the validation in two models for Type 2 Diabetes (T2D)–the ZDF rat and db/db mouse. We present a full validation and comparison of two animal models for Duchenne Muscular Dystrophy (DMD): the mdx mouse and GRMD dog. We show that there are significant differences between the mdx mouse and the GRMD dog, the latter mimicking the human epidemiological, SNH, and histological aspects to a greater extent than the mouse despite the overall lack of published data. This does not alter our adherence to PLOS ONE policies on sharing data and materials

Conclusions
SYMPTOMATOLOGY AND NATURAL HISTORY VALIDATION
GENETIC VALIDATION
BIOCHEMICAL VALIDATION
ENDPOINT VALIDATION
Limitations
Findings
31.4 Background Model
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call